Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
2.
Acta Biomater ; 180: 18-45, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38641182

RESUMEN

Bacterial extracellular vesicles (BEVs) are naturally occurring bioactive membrane-bound nanoparticles released by both gram-negative and gram-positive bacterial species, exhibiting a multifaceted role in mediating host-microbe interactions across various physiological conditions. Increasing evidence supports BEVs as essential mediators of cell-to-cell communicaiton, influencing bacterial pathogenicity, disease mechanisms, and modulating the host immune response. However, the extent to which these BEV-mediated actions can be leveraged to predict disease onset, guide treatment strategies, and determine clinical outcomes remains uncertain, particularly in terms of their clinical translation potentials. This review briefly describes BEV biogenesis and their internalisation by recipient cells and summarises methods for isolation and characterization, essential for understanding their composition and cargo. Further, it discusses the potential of biofluid-associated BEVs as biomarkers for various diseases, spanning both cancer and non-cancerous conditions. Following this, we outline the ongoing human clinical trials of using BEVs for vaccine development. In addition to disease diagnostics, this review explores the emerging research of using natural or engineered BEVs as smart nanomaterials for applications in anti-cancer therapy and bone regeneration. This discussion extends to key factors for unlocking the clinical potential of BEVs, such as standardization of BEV isolation and characterisation, as well as other hurdles in translating these findings to the clinical setting. We propose that addressing these hurdles through collaborative research efforts and well-designed clinical trials holds the key to fully harnessing the clinical potential of BEVs. As this field advances, this review suggests that BEV-based nanomedicine has the potential to revolutionize disease management, paving the way for innovative diagnosis, therapeutics, and personalized medicine approaches. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) from both host cells and bacteria serve as multifunctional biomaterials and are emerging in the fields of biomedicine, bioengineering, and biomaterials. However, the majority of current studies focus on host-derived EVs, leaving a gap in comprehensive research on bacteria-derived EVs (BEVs). Although BEVs offer an attractive option as nanomaterials for drug delivery systems, their unique nanostructure and easy-to-modify functions make them a potential method for disease diagnosis and treatment as well as vaccine development. Our work among the pioneering studies investigating the potential of BEVs as natural nanobiomaterials plays a crucial role in both understanding the development of diseases and therapeutic interventions.


Asunto(s)
Vesículas Extracelulares , Nanoestructuras , Vesículas Extracelulares/metabolismo , Humanos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Animales , Bacterias/metabolismo , Neoplasias/terapia , Neoplasias/patología
3.
Biomater Adv ; 158: 213770, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38242057

RESUMEN

Recent research indicates that combining 3D bioprinting and small extracellular vesicles (sEVs) offers a promising 'cell-free' regenerative medicine approach for various tissue engineering applications. Nonetheless, the majority of existing research has focused on bioprinting of sEVs sourced from cell lines. There remains a notable gap in research regarding the bioprinting of sEVs derived from primary human periodontal cells and their potential impact on ligamentous and osteogenic differentiation. Here, we investigated the effect of 3D bioprinted periodontal cell sEVs constructs on the differentiation potential of human buccal fat pad-derived mesenchymal stromal cells (hBFP-MSCs). Periodontal cell-derived sEVs were enriched by size exclusion chromatography (SEC) with particle-shaped morphology, and characterized by being smaller than 200 nm in size and CD9/CD63/CD81 positive, from primary human periodontal ligament cells (hPDLCs) and human gingival fibroblasts (hGFs). The sEVs were then 3D bioprinted in 10 % gelatin methacryloyl (GelMA) via microextrusion bioprinting. Release of sEVs from bioprinted constructs was determined by DiO-labelling and confocal imaging, and CD9 ELISA. Attachment and ligament/osteogenic/cementogenic differentiation of hBFP-MSCs was assessed on bioprinted GelMA, without and with sEVs (GelMA/hPDLCs-sEVs and GelMA/hGFs-sEVs), scaffolds. hBFP-MSCs seeded on the bioprinted sEVs constructs spread well with significantly enhanced focal adhesion, mechanotransduction associated gene expression, and ligament and osteogenesis/cementogenesis differentiation markers in GelMA/hPDLCs-sEVs, compared to GelMA/hGFs-sEVs and GelMA groups. A 2-week osteogenic and ligamentous differentiation showed enhanced ALP staining, calcium formation and toluidine blue stained cells in hBFP-MSCs on bioprinted GelMA/hPDLCs-sEVs constructs compared to the other two groups. The proof-of-concept data from this study supports the notion that 3D bioprinted GelMA/hPDLCs-sEVs scaffolds promote cell attachment, as well as ligamentous, osteogenic and cementogenic differentiation, of hBFP-MSCs in vitro.


Asunto(s)
Células Madre Mesenquimatosas , Andamios del Tejido , Humanos , Andamios del Tejido/química , Osteogénesis , Mecanotransducción Celular , Ingeniería de Tejidos/métodos
4.
J Periodontal Res ; 58(6): 1188-1200, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37605485

RESUMEN

OBJECTIVE: To enrich and compare three extracellular vesicles-EV subtypes (apoptotic bodies, microvesicles and small EV) from three periodontal cells (periodontal ligament cells-PDLCs, alveolar bone-derived osteoblasts-OBs and gingival fibroblasts-GFs), and assess uptake and cell function changes in buccal fat pad-derived mesenchymal stromal cells (BFP-MSCs). BACKGROUND: Periodontal cells such as PDLCs, OBs and GFs have the potential to enhance bone and periodontal regeneration, but face significant challenges, such as the regulatory and cost implications of in vitro cell culture and storage. To address these challenges, it is important to explore alternative 'cell-free' strategies, such as extracellular vesicles which have emerged as promising tools in regenerative medicine, to facilitate osteogenic differentiation and bone regeneration. METHODS AND MATERIALS: Serial centrifuges at 2600 and 16 000 g were used to isolate apoptotic bodies and microvesicles respectively. Small EV-sEV was enriched by our in-house size exclusion chromatography (SEC). The cellular uptake, proliferation, migration and osteogenic/adipogenic differentiation genes were analysed after EVs uptake in BFP-MSCs. RESULTS: Three EV subtypes were enriched and characterised by morphology, particle size and EV-associated protein expression-CD9. Cellular uptake of the three EVs subtypes was observed in BFP-MSCs for up to 7 days. sEV from the three periodontal cells promoted proliferation, migration and osteogenic gene expression. hOBs-sEV showed superior levels of osteogenesis markers compared to that hPDLCs-sEV and hGFs-sEV, while hOBs-16k EV promoted adipogenic gene expression compared to that from hPDLCs and hGFs. CONCLUSIONS: Our proof-of-concept data demonstrate that hOBs-sEV might be an alternative cell-free therapeutic for bone tissue engineering.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Osteogénesis , Diferenciación Celular , Vesículas Extracelulares/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas
5.
Nanomaterials (Basel) ; 13(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37446538

RESUMEN

Zinc (Zn) as a biodegradable metal has attracted research interest for bone reconstruction, with the aim of eliminating the need for a second removal surgery and minimizing the implant-to-bone transfer of stress-shielding to maintain bone regeneration. In addition, Zn has been shown to have antibacterial properties, particularly against Gram-negative bacteria, and is often used as a surface coating to inhibit bacterial growth and biofilm formation. However, the antibacterial property of Zn is still suboptimal in part due to low Zn ion release during degradation that has to be further improved in order to meet clinical requirements. This work aims to perform an innovative one-step surface modification using a nitric acid treatment to accelerate Zn ion release by increasing surface roughness, thereby endowing an effective antimicrobial property and biofilm formation inhibition. The antibacterial performance against Staphylococci aureus was evaluated by assessing biofilm formation and adhesion using quantitative assays. The surface roughness of acid-treated Zn (Ra ~ 30 nm) was significantly higher than polished Zn (Ra ~ 3 nm) and corresponded with the marked inhibition of bacterial biofilm, and this is likely due to the increased surface contact area and Zn ion accumulation. Overall, surface modification due to nitric acid etching appears to be an effective technique that can produce unique morphological surface structures and enhance the antibacterial properties of biodegradable Zn-based materials, thus increasing the translation potential toward multiple biomedical applications.

6.
Tissue Eng Part C Methods ; 29(7): 298-306, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37358387

RESUMEN

This cross-sectional pilot study explored extracellular vesicle (EV)-derived gene expression of markers for bone turnover and pro-inflammatory cytokines in periodontal disease. Whole unstimulated saliva was collected from 52 participants (18 healthy, 13 gingivitis, and 21 stages III/IV periodontitis), from which salivary small extracellular vesicles (sEVs) were enriched using the size-exclusion chromatography method, and characterized by morphology, EV-protein, and size distribution, using transmission electron microscopy (TEM), enzyme-linked immunosorbent assay (ELISA), and Nanoparticle Tracking Analysis (NTA), respectively. Bone turnover markers and pro-inflammatory cytokines in salivary sEVs were evaluated using reverse transcription PCR. Salivary sEVs morphology, mode, size distribution, and particle concentration were comparable between healthy, gingivitis, and periodontitis patients. The CD9+ subpopulation was significantly higher in periodontitis-derived salivary sEVs compared with healthy. The detection of sEVs mRNA for osterix and tumor necrosis factor-alpha was significantly decreased and increased, respectively, in periodontitis compared with healthy controls, with good discriminatory power for periodontitis diagnosis (area under the curve >0.72). This pilot study demonstrated that salivary sEVs mRNAs may serve as a potential noninvasive biomarker source for periodontitis diagnosis.


Asunto(s)
Gingivitis , Periodontitis , Humanos , Factor de Necrosis Tumoral alfa/análisis , Proyectos Piloto , ARN Mensajero/genética , Estudios Transversales , Citocinas
7.
Trends Pharmacol Sci ; 44(6): 324-334, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36805364

RESUMEN

Adenosine-lidocaine-magnesium (ALM) mixture is a cardioplegic agent that improves survivability undisputedly in rodents, but not swine, models of hemorrhagic shock. However, despite protection from comorbid coagulopathy being the one common effect reported in both models, the underlying prothrombotic mechanism for ALM has not been fully elucidated. Here, we undertook a component-based approach focusing on individual drugs in the mixture to elaborate on the protective mechanism against coagulopathy within the frames of adenosine signaling and metabolic pathways. Additionally, the translational potential of small and large animal models of hemorrhagic shock for human survival is critically appraised, owing to substantial quantitative/qualitative differences between humans and rodents, particularly regarding the genetics of G protein-coupled receptors (GPCRs) interacting with ALM's constituents.


Asunto(s)
Choque Hemorrágico , Humanos , Porcinos , Animales , Magnesio/farmacología , Adenosina , Lidocaína/farmacología , Modelos Animales de Enfermedad
8.
J Periodontol ; 94(1): 77-87, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35771077

RESUMEN

BACKGROUND: This prospective cohort study aimed to evaluate the antibody responses in non-invasive gingival crevicular fluid (GCF) and unstimulated whole saliva to the SARS-CoV-2 Spike unit 1 receptor-binding domain (S1-RBD) protein following administration of the mRNA BNT162b2 vaccine. METHODS: This longitudinal study recruited 37 participants with no prior COVID-19 exposure (eight people recruited prior to the COVID-19 pandemic - labeled pre-COVID, 16 vaccinated and 13 non-vaccinated participants). An enzyme-linked immunosorbent assay (ELISA) was used to determine antibody levels against S1-RBD in saliva (n=90) and GCF (n=80) samples obtained at 1 and 3 weeks after dose 1, and 3 days, 7 days, and 3 weeks after dose 2. To determine previous SARS-CoV-2 infection status, anti-nucleocapsid (N) Ig levels were determined in samples from the pre-COVID (saliva as reference), non-vaccinated (saliva and GCF), and vaccinated (saliva and GCF) participants at 1-week post-dose 1 using ELISA. RESULTS: Salivary levels of anti-N antibodies measured in samples from vaccinated and nonvaccinated participants were comparable to those in pre-COVID saliva samples collected between October 2018 and September 2019, thus confirming that all study participants had no prior SARS-CoV-2 infection. Overall, the levels of anti-S1-RBD antibodies peaked at 3 weeks after dose 2 in both saliva and GCF for all three immunoglobulin isotypes. Notably, the concentration of anti-S1-RBD antibodies in GCF was significantly higher than in saliva at all time points. CONCLUSION: This study establishes GCF and saliva as viable alternative non-invasive sources to monitor levels of antibodies following vaccination, with GCF demonstrating feasibility as a biofluid source for the detection of antibodies against SARS-CoV-2 S1-RBD antigen.


Asunto(s)
COVID-19 , Líquido del Surco Gingival , Humanos , Líquido del Surco Gingival/química , Vacuna BNT162 , COVID-19/prevención & control , COVID-19/metabolismo , Formación de Anticuerpos , Pandemias , Estudios Longitudinales , Estudios Prospectivos , SARS-CoV-2 , Vacunas de ARNm
9.
Cardiovasc Ther ; 2022: 5299370, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36262119

RESUMEN

Background and Aims: The nacht domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is upregulated in human abdominal aortic aneurysm (AAA), but its pathogenic role is unclear. The aims of this study were firstly to examine whether the inflammasome was upregulated in a mouse model of AAA and secondly to test whether the inflammasome inhibitor colchicine limited AAA growth. Methods: AAA was induced in eight-week-old male C57BL6/J mice with topical application of elastase to the infrarenal aorta and oral 3-aminopropionitrile (E-BAPN). For aim one, inflammasome activation, abdominal aortic diameter, and rupture were compared between mice with AAA and sham controls. For aim two, 3 weeks after AAA induction, mice were randomly allocated to receive colchicine (n = 28, 0.2 mg/kg/d) or vehicle control (n = 29). The primary outcome was the rate of maximum aortic diameter increase measured by ultrasound over 13 weeks. Results: There was upregulation of NLRP3 markers interleukin- (IL-) 1ß (median, IQR; 15.67, 7.11-22.60 pg/mg protein versus 6.87, 4.54-11.60 pg/mg protein, p = .048) and caspase-1 (109, 83-155 relative luminosity units (RLU) versus 45, 38-65 RLU, p < .001) in AAA samples compared to controls. Aortic diameter increase over 80 days (mean difference, MD, 4.3 mm, 95% CI 3.3, 5.3, p < .001) was significantly greater in mice in which aneurysms were induced compared to sham controls. Colchicine did not significantly limit aortic diameter increase over 80 days (MD -0.1 mm, 95% CI -1.1, 0.86, p = .922). Conclusions: The inflammasome was activated in this mouse model of AAA; however, daily oral administration of colchicine did not limit AAA growth.


Asunto(s)
Aneurisma de la Aorta Abdominal , Animales , Masculino , Ratones , Aminopropionitrilo , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Caspasas , Colchicina/farmacología , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Leucina , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Elastasa Pancreática
10.
RSC Adv ; 12(38): 24849-24856, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36128389

RESUMEN

Sensitive detection of immunoglobulin antibodies against SARS-CoV-2 during the COVID-19 pandemic is critical to monitor the adaptive immune response after BNT162b2 mRNA vaccination. Currently employed binding antibody detection tests using 2D microplate-based enzyme-linked immunosorbent assays (ELISA) are limited by the degree of sensitivity. In this study, a 3D antibody test was developed by immobilizing the receptor-binding domain on Spike subunit 1 (S1-RBD) of SARS-CoV-2 onto engineered melt electrowritten (MEW) poly(ε-caprolactone) (PCL) scaffolds (pore: 500 µm, fiber diameter: 17 µm) using carbodiimide crosslinker chemistry. Protein immobilization was confirmed using X-ray photoelectron spectroscopy (XPS) by the presence of peaks corresponding with nitrogen. Self-developed indirect ELISA was performed to assess the functionality of the 3D platform in comparison with a standard 2D tissue culture plate (TCP) system, using whole unstimulated saliva samples from 14 non-vaccinated and 20 vaccinated participants (1- and 3- weeks post-dose 1; 3 days, 1 week and 3 weeks post-dose 2) without prior SARS-CoV-2 infection. The three-dimensional S1-RBD PCL scaffolds, while demonstrating a kinetic trend comparable to 2D TCP, exhibited significantly higher sensitivity and detection levels for all three immunoglobulins assayed (IgG, IgM, and IgA). These novel findings highlight the potential of MEW PCL constructs in the development of improved low-cost, point-of-care, and self-assessing diagnostic platforms for the detection and monitoring of SARS-CoV-2 antibodies.

11.
ACS Appl Mater Interfaces ; 14(19): 22554-22569, 2022 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-35533291

RESUMEN

Zinc (Zn) has recently been identified as an auspicious biodegradable metal for medical implants and devices due to its tunable mechanical properties and good biocompatibility. However, the slow corrosion rate of Zn in a physiological environment does not meet the requirements for biodegradable implants, hindering its clinical translation. The present study aimed to accelerate the corrosion rate of pure Zn by utilizing acid etching to roughen the surface and increase the substrate surface area. The effects of acid etching on surface morphology, surface roughness, tensile properties, hardness, electrochemical corrosion and degradation behavior, cytocompatibility, direct cell attachment, and biofilm formation were investigated. Interestingly, acid-treated Zn showed an exceptionally high rate of corrosion (∼226-125 µm/year) compared to untreated Zn (∼62 µm/year), attributed to the increased surface roughness (Ra ∼ 1.12 µm) of acid-etched samples. Immersion tests in Hank's solution revealed that acid etching accelerated the degradation rate of Zn samples. In vitro, MC3T3-E1 cell lines in 50 and 25% conditioned media extracts of treated samples showed good cytocompatibility. Reduced bacterial adhesion, biofilm formation, and dispersion were observed for Staphylococci aureus biofilms cultured on acid-etched pure Zn substrates. These results suggest that the surface modification of biodegradable pure Zn metals by acid etching markedly increases the translation potential of zinc for various biomedical applications.


Asunto(s)
Aleaciones , Zinc , Implantes Absorbibles , Aleaciones/química , Antibacterianos/farmacología , Materiales Biocompatibles , Corrosión , Ensayo de Materiales , Zinc/química
12.
Cytokine Growth Factor Rev ; 64: 7-11, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35115234

RESUMEN

Osteoporosis results from dysregulated bone remodeling with increased osteoclast-mediated destruction of bones. We have recently shown in vitro the truncated tryptophanyl-tRNA synthetase (mini-TrpRS)-dependent action of interferon-gamma (IFN-γ) to promote myeloid lineage multinucleation, a fundamental step in the osteoclast formation. In particular, we found that IFN-γ readily induced monocyte aggregation leading to multinuclear giant cell formation that paralleled marked upregulation of mini-TrpRS. However, blockade of mini-TrpRS with its cognate amino acid and decoy substrate D-Tryptophan prevented mini-TrpRS signaling, and markedly reduced the aggregation of monocytes and multinucleation in the presence of IFN. The cell signaling mechanism executed by mini-TrpRS appears inevitably in any inflammatory environment that involves IFN-γ with outcomes depending on the cell type involved. Here, we elaborate on these findings and discuss the potential role of the IFN-γ/mini-TrpRS signaling axis in osteoporosis pathophysiology, which may eventually materialize in a novel therapeutic perspective for this disease.


Asunto(s)
Osteoporosis , Triptófano-ARNt Ligasa , Humanos , Interferón gamma , Osteoporosis/tratamiento farmacológico , Unión Proteica , Transducción de Señal , Triptófano-ARNt Ligasa/química , Triptófano-ARNt Ligasa/genética , Triptófano-ARNt Ligasa/metabolismo
13.
Trends Cardiovasc Med ; 32(3): 138-142, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33571665

RESUMEN

Atherosclerosis demonstrates an increased rate of vascular smooth muscle cells (VSMC) plasticity characterized by switching from the differentiated contractile phenotype to a de-differentiated synthetic state. In healthy blood vessels, phenotypic switching represents a fundamental property of VSMC in maintaining vascular homeostasis. However, in atherosclerosis, it is an initial and necessary step in VSMC-derived foam cell formation. These foam cells play a decisive role in atherosclerosis progression since approximately half of all the foam cells are of VSMC origin. Our recent work showed that interferon-gamma (IFN-γ), a primary inflammatory cytokine in progressive atherosclerosis, mediates VSMC phenotype switching exclusively through upregulating mini-tryptophanyl-tRNA synthetase (mini-TrpRS). Here, we discuss the pro-atherosclerotic implication of this phenomenon that inevitably occurs in the context of a more complex regulation mediated by IFN-γ. An emerging therapeutic option for patients with progressive atherosclerosis is also discussed.


Asunto(s)
Aterosclerosis , Citocinas , Aterosclerosis/terapia , Humanos , Músculo Liso Vascular , Miocitos del Músculo Liso , Fenotipo , Transducción de Señal
14.
Sci Rep ; 11(1): 17451, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465809

RESUMEN

Inflammation, vascular smooth muscle cell apoptosis and oxidative stress are believed to play important roles in abdominal aortic aneurysm (AAA) pathogenesis. Human kallistatin (KAL; gene SERPINA4) is a serine proteinase inhibitor previously shown to inhibit inflammation, apoptosis and oxidative stress. The aim of this study was to investigate the role of KAL in AAA through studies in experimental mouse models and patients. Serum KAL concentration was negatively associated with the diagnosis and growth of human AAA. Transgenic overexpression of the human KAL gene (KS-Tg) or administration of recombinant human KAL (rhKAL) inhibited AAA in the calcium phosphate (CaPO4) and subcutaneous angiotensin II (AngII) infusion mouse models. Upregulation of KAL in both models resulted in reduction in the severity of aortic elastin degradation, reduced markers of oxidative stress and less vascular smooth muscle apoptosis within the aorta. Administration of rhKAL to vascular smooth muscle cells incubated in the presence of AngII or in human AAA thrombus-conditioned media reduced apoptosis and downregulated markers of oxidative stress. These effects of KAL were associated with upregulation of Sirtuin 1 activity within the aortas of both KS-Tg mice and rodents receiving rhKAL. These results suggest KAL-Sirtuin 1 signalling limits aortic wall remodelling and aneurysm development through reductions in oxidative stress and vascular smooth muscle cell apoptosis. Upregulating KAL may be a novel therapeutic strategy for AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Apoptosis , Aterosclerosis/prevención & control , Modelos Animales de Enfermedad , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Serpinas/administración & dosificación , Anciano , Anciano de 80 o más Años , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Serpinas/sangre
15.
Cytokine ; 142: 155486, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33721618

RESUMEN

Truncated tryptophanyl-tRNA synthetase (mini-TrpRS), like any other aminoacyl-tRNA synthetases, canonically functions as a protein synthesis enzyme. Here we provide evidence for an additional signaling role of mini-TrpRS in the formation of monocyte-derived multinuclear giant cells (MGCs). Interferon-gamma (IFNγ) readily induced monocyte aggregation leading to MGC formation with paralleled marked upregulation of mini-TrpRS. Small interfering (si)RNA, targeting mini-TrpRS in the presence of IFNγ prevented monocyte aggregation. Moreover, blockade of mini-TrpRS, either by siRNA, or the cognate amino acid and decoy substrate D-Tryptophan to prevent mini-TrpRS signaling, resulted in a marked reduction in expression of the purinergic receptor P2X 7 (P2RX7) in monocytes activated by IFNγ. Our findings identify mini-TrpRS as a critical signaling molecule in a mechanism by which IFNγ initiates monocyte-derived giant cell formation.


Asunto(s)
Células Gigantes/citología , Células Gigantes/enzimología , Interferón gamma/farmacología , Monocitos/citología , Triptófano-ARNt Ligasa/metabolismo , Agregación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Humanos , Modelos Biológicos , Receptores Purinérgicos/metabolismo , Transducción de Señal/efectos de los fármacos , Células THP-1 , Regulación hacia Arriba/efectos de los fármacos
16.
J Am Heart Assoc ; 10(5): e019372, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33599139

RESUMEN

Background Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. The kinin B2 receptor agonist, bradykinin, has been implicated in AAA pathogenesis through promoting inflammation. Bradykinin is generated from high- and low-molecular-weight kininogen by the serine protease kallikrein-1. The aims of this study were first to examine the effect of neutralizing kallikrein-1 on AAA development in a mouse model and second to test how blocking kallikrein-1 affected cyclooxygenase-2 and prostaglandin E2 in human AAA explants. Methods and Results Neutralization of kallikrein-1 in apolipoprotein E-deficient (ApoE-/-) mice via administration of a blocking antibody inhibited suprarenal aorta expansion in response to angiotensin (Ang) II infusion. Kallikrein-1 neutralization decreased suprarenal aorta concentrations of bradykinin and prostaglandin E2 and reduced cyclooxygenase-2 activity. Kallikrein-1 neutralization also decreased protein kinase B and extracellular signal-regulated kinase 1/2 phosphorylation and reduced levels of active matrix metalloproteinase 2 and matrix metalloproteinase 9. Kallikrein-1 blocking antibody reduced levels of cyclooxygenase-2 and secretion of prostaglandin E2 and active matrix metalloproteinase 2 and matrix metalloproteinase 9 from human AAA explants and vascular smooth muscle cells exposed to activated neutrophils. Conclusions These findings suggest that kallikrein-1 neutralization could be a treatment target for AAA.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/terapia , Dinoprostona/metabolismo , Músculo Liso Vascular/patología , Calicreínas de Tejido/antagonistas & inhibidores , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Biopsia , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo
17.
Clin Sci (Lond) ; 134(18): 2521-2534, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32936248

RESUMEN

Vitamin D deficiency has been associated with human abdominal aortic aneurysm (AAA); however, its role in AAA pathogenesis is unclear. The aim of the present study was to investigate the effect of vitamin D deficiency on AAA development and examine if administering cholecalciferol (CCF) could limit growth of established AAA within the angiotensin-II (AngII) infused apolipoprotein E-deficient mouse model. Mice were rendered vitamin D deficiency through dietary restriction and during AngII infusion developed larger AAAs as assessed by ultrasound and ex vivo morphometry that ruptured more commonly (48% vs. 19%; P=0.028) than controls. Vitamin D deficiency was associated with increased aortic expression of osteopontin and matrix metalloproteinase-2 and -9 than controls. CCF administration to mice with established aortic aneurysms limited AAA growth as assessed by ultrasound (P<0.001) and ex vivo morphometry (P=0.036) and reduced rupture rate (8% vs. 46%; P=0.031). This effect was associated with up-regulation of circulating and aortic sclerostin. Incubation of human aortic smooth muscle cells with 1,25-dihyroxyvitamin D3 (the active metabolite of vitamin D) for 48 h induced up-regulation of sclerostin (P<0.001) and changed the expression of a range of other genes important in extracellular matrix remodeling. The present study suggests that vitamin D deficiency promotes development of large rupture-prone aortic aneurysms in an experimental model. CCF administration limited both growth and rupture of established aneurysms. These effects of vitamin D appeared to be mediated via changes in genes involved in extracellular matrix remodeling, particularly sclerostin.


Asunto(s)
Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Aneurisma de la Aorta Abdominal/etiología , Rotura de la Aorta/tratamiento farmacológico , Rotura de la Aorta/etiología , Colecalciferol/uso terapéutico , Suplementos Dietéticos , Progresión de la Enfermedad , Deficiencia de Vitamina D/complicaciones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Aneurisma de la Aorta Abdominal/fisiopatología , Rotura de la Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Presión Sanguínea/efectos de los fármacos , Restricción Calórica , Colecalciferol/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Deficiencia de Vitamina D/fisiopatología
18.
Eur J Vasc Endovasc Surg ; 60(3): 452-460, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32703634

RESUMEN

OBJECTIVE: Experimental studies suggest that fenofibrate prevents abdominal aortic aneurysm (AAA) development by lowering aortic osteopontin (OPN) concentration and reducing the number of macrophages infiltrating the aortic wall. The current study examined the effects of a short course of fenofibrate on AAA pathology in people with large AAAs awaiting aortic repair. METHODS: This randomised double blind parallel trial included male and female participants aged ≥ 60 years who had an asymptomatic AAA measuring ≥ 50 mm and were scheduled to undergo open AAA repair. Participants were allocated to fenofibrate (145 mg/day) or matching placebo for at least two weeks before elective AAA repair. Blood samples were collected at recruitment and immediately prior to surgery. AAA biopsies were obtained during aortic surgery. The primary outcomes were (1) AAA OPN concentration; (2) serum OPN concentration; and (3) number of AAA macrophages. Exploratory outcomes included circulating and aortic concentrations of other proteins previously associated with AAA. Outcomes assessed at a single time point were compared using logistic regression. Longitudinal outcomes were compared using linear mixed effects models. RESULTS: Forty-three participants were randomised. After three withdrawals, 40 were followed until the time of surgery (21 allocated fenofibrate and 19 allocated placebo). As expected, serum triglycerides reduced significantly from recruitment to the time of surgery in participants allocated fenofibrate. No differences in any of the primary and exploratory outcomes were observed between groups. CONCLUSION: A short course of 145 mg of fenofibrate/day did not lower concentrations of OPN or aortic macrophage density in people with large AAAs.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/cirugía , Aneurisma de la Aorta Abdominal/terapia , Fenofibrato/administración & dosificación , Procedimientos Quirúrgicos Vasculares , Anciano , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/patología , Biomarcadores/sangre , Progresión de la Enfermedad , Método Doble Ciego , Esquema de Medicación , Femenino , Fenofibrato/efectos adversos , Humanos , Macrófagos/patología , Masculino , Persona de Mediana Edad , Osteopontina/sangre , Queensland , Factores de Tiempo , Resultado del Tratamiento , Triglicéridos/sangre , Remodelación Vascular/efectos de los fármacos , Procedimientos Quirúrgicos Vasculares/efectos adversos
19.
Clin Sci (Lond) ; 134(9): 1049-1061, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32309850

RESUMEN

Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Factor XII/antagonistas & inhibidores , Proteína ADAM17/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/metabolismo , Modelos Animales de Enfermedad , Factor XII/metabolismo , Ratones
20.
Cytokine ; 127: 154940, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31786502

RESUMEN

Phenotypic modulation of vascular smooth muscle cells (AoSMCs) between quiescent 'contractile' and active 'synthetic' states is crucial in response to normal stimuli and pathological stressors. Previous studies have revealed the ability of interferon gamma (IFN-γ) to activate and promote a synthetic phenotype in AoSMCs that parallels marked up-regulation of truncated tryptophanyl-tRNA synthetase (mini-TrpRS). Here we provide evidence to support an essential dependency of IFN-γ-induced activation and synthetic phenotype in AoSMC on mini-TrpRS. This is based upon change in AoSMC morphology from epithelioid (active synthetic) to spindle-shaped (quiescent contractile) cells and expression of proteins and genes important in mediating or regulating contractile function of AoSMCs, following blockade of mini-TrpRS induced by IFN-γ, via targeted siRNA or the decoy cognate amino acid D-Tryptophan.


Asunto(s)
Interferón gamma/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Receptores Adrenérgicos beta 2/genética , Triptófano-ARNt Ligasa/genética , Proteínas de Unión al Calcio , Calmodulina , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Miocitos del Músculo Liso/metabolismo , Quinasa de Cadena Ligera de Miosina , Proteínas Nucleares , Fenotipo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transactivadores , Triptófano/metabolismo , Triptófano/farmacología , Triptófano-ARNt Ligasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...